1
|
Li Y, Sun Y, Yu K, Li Z, Miao H, Xiao W. Keratin: A potential driver of tumor metastasis. Int J Biol Macromol 2025; 307:141752. [PMID: 40049479 DOI: 10.1016/j.ijbiomac.2025.141752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Keratins, as essential components of intermediate filaments in epithelial cells, play a crucial role in maintaining cell structure and function. In various malignant epithelial tumors, abnormal keratin expression is frequently observed and serves not only as a diagnostic marker but also closely correlates with tumor progression. Extensive research has demonstrated that keratins are pivotal in multiple stages of tumor metastasis, including responding to mechanical forces, evading the immune system, reprogramming metabolism, promoting angiogenesis, and resisting apoptosis. Here we emphasize that keratins significantly enhance the migratory and invasive capabilities of tumor cells, making them critical drivers of tumor metastasis. These findings highlight the importance of targeting keratins as a strategic approach to combat tumor metastasis, thereby advancing our understanding of their role in cancer progression and offering new therapeutic opportunities.
Collapse
Affiliation(s)
- Yuening Li
- Army Medical University, Chongqing, China
| | - Yiming Sun
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Kun Yu
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhixi Li
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Jinfeng Laboratory, Chongqing, China.
| | - Weidong Xiao
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
2
|
Nyström JH, Heikkilä TRH, Thapa K, Pulli I, Törnquist K, Toivola DM. Colonocyte keratins stabilize mitochondria and contribute to mitochondrial energy metabolism. Am J Physiol Gastrointest Liver Physiol 2024; 327:G438-G453. [PMID: 38860856 PMCID: PMC11427106 DOI: 10.1152/ajpgi.00220.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Keratin intermediate filaments form dynamic filamentous networks, which provide mechanical stability, scaffolding, and protection against stress to epithelial cells. Keratins and other intermediate filaments have been increasingly linked to the regulation of mitochondrial function and homeostasis in different tissues and cell types. While deletion of keratin 8 (K8-/-) in mouse colon elicits a colitis-like phenotype, epithelial hyperproliferation, and blunted mitochondrial ketogenesis, the role of K8 in colonocyte mitochondrial function and energy metabolism is unknown. We used two K8 knockout mouse models and CRISPR/Cas9 K8-/- colorectal adenocarcinoma Caco-2 cells to answer this question. The results show that K8-/- colonocyte mitochondria in vivo are smaller and rounder and that mitochondrial motility is increased in K8-/- Caco-2 cells. Furthermore, K8-/- Caco-2 cells displayed diminished mitochondrial respiration and decreased mitochondrial membrane potential compared with controls, whereas glycolysis was not affected. The levels of mitochondrial respiratory chain complex proteins and mitochondrial regulatory proteins mitofusin-2 and prohibitin were decreased both in vitro in K8-/- Caco-2 cells and in vivo in K8-/- mouse colonocytes, and reexpression of K8 into K8-/- Caco-2 cells normalizes the mitofusin-2 levels. Mitochondrial Ca2+ is an important regulator of mitochondrial energy metabolism and homeostasis, and Caco-2 cells lacking K8 displayed decreased levels and altered dynamics of mitochondrial matrix and cytoplasmic Ca2+. In summary, these novel findings attribute an important role for colonocyte K8 in stabilizing mitochondrial shape and movement and maintaining mitochondrial respiration and Ca2+ signaling. Further, how these metabolically compromised colonocytes are capable of hyperproliferating presents an intriguing question for future studies.NEW & NOTEWORTHY In this study, we show that colonocyte intermediate filament protein keratin 8 is important for stabilizing mitochondria and maintaining mitochondrial energy metabolism, as keratin 8-deficient colonocytes display smaller, rounder, and more motile mitochondria, diminished mitochondrial respiration, and altered Ca2+ dynamics. Changes in fusion-regulating proteins are rescued with reexpression of keratin 8. These alterations in colonocyte mitochondrial homeostasis contribute to keratin 8-associated colitis pathophysiology.
Collapse
Affiliation(s)
- Joel H Nyström
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Taina R H Heikkilä
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Keshav Thapa
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ilari Pulli
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Kid Törnquist
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
3
|
Baghestani S, Haldin C, Kosijer P, Alam CM, Toivola DM. β-Cell keratin 8 maintains islet mechanical integrity, mitochondrial ultrastructure, and β-cell glucose transporter 2 plasma membrane targeting. Am J Physiol Cell Physiol 2024; 327:C462-C476. [PMID: 38912736 DOI: 10.1152/ajpcell.00123.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024]
Abstract
Islet β-cell dysfunction is an underlying factor for type I diabetes (T1D) development. Insulin sensing and secretion are tightly regulated in β-cells at multiple subcellular levels. The epithelial intermediate filament (IF) protein keratin (K) 8 is the main β-cell keratin, constituting the filament network with K18. To identify the cell-autonomous functions of K8 in β-cells, mice with targeted deletion of β-cell K8 (K8flox/flox; Ins-Cre) were analyzed for islet morphology, ultrastructure, and integrity, as well as blood glucose regulation and streptozotocin (STZ)-induced diabetes development. Glucose transporter 2 (GLUT2) localization was studied in β-cells in vivo and in MIN6 cells with intact or disrupted K8/K18 filaments. Loss of β-cell K8 leads to a major reduction in K18. Islets without β-cell K8 are more fragile, and these β-cells display disjointed plasma membrane organization with less membranous E-cadherin and smaller mitochondria with diffuse cristae. Lack of β-cell K8 also leads to a reduced glucose-stimulated insulin secretion (GSIS) response in vivo, despite undisturbed systemic blood glucose regulation. K8flox/flox, Ins-Cre mice have a decreased sensitivity to STZ compared with K8 wild-type mice, which is in line with decreased membranous GLUT2 expression observed in vivo, as GLUT2 is required for STZ uptake in β-cells. In vitro, MIN6 cell plasma membrane GLUT2 is rescued in cells overexpressing K8/K18 filaments but mistargeted in cells with disrupted K8/K18 filaments. β-Cell K8 is required for islet and β-cell structural integrity, normal mitochondrial morphology, and GLUT2 plasma membrane targeting, and has implications on STZ sensitivity as well as systemic insulin responses.NEW & NOTEWORTHY Keratin 8 is the main cytoskeletal protein in the cytoplasmic intermediate filament network in β-cells. Here for the first time, we assessed the β-cell autonomous mechanical and nonmechanical roles of keratin 8 in β-cell function. We demonstrated the importance of keratin 8 in islet and β-cell structural integrity, maintaining mitochondrial morphology and GLUT2 plasma membrane targeting.
Collapse
Affiliation(s)
- Sarah Baghestani
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Caroline Haldin
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Petar Kosijer
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Catharina M Alam
- School of Applied Sciences, Edinburgh Napier University, Edinburg, United Kingdom
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
4
|
Meng X, Zhou Y, Xu L, Hu L, Wang C, Tian X, Zhang X, Hao Y, Cheng B, Ma J, Wang L, Liu J, Xie R. O-GlcNAcylation Facilitates the Interaction between Keratin 18 and Isocitrate Dehydrogenases and Potentially Influencing Cholangiocarcinoma Progression. ACS CENTRAL SCIENCE 2024; 10:1065-1083. [PMID: 38799671 PMCID: PMC11117311 DOI: 10.1021/acscentsci.4c00163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/06/2024] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
Glycosylation plays a pivotal role in the intricate landscape of human cholangiocarcinoma (CCA), actively participating in key pathophysiological processes driving tumor progression. Among the various glycosylation modifications, O-linked β-N-acetyl-glucosamine modification (O-GlcNAcylation) emerges as a dynamic regulator influencing diverse tumor-associated biological activities. In this study, we employed a state-of-the-art chemical proteomic approach to analyze intact glycopeptides, unveiling the critical role of O-GlcNAcylation in orchestrating Keratin 18 (K18) and its interplay with tricarboxylic acid (TCA) cycle enzymes, specifically isocitrate dehydrogenases (IDHs), to propel CCA progression. Our findings shed light on the mechanistic intricacies of O-GlcNAcylation, revealing that site-specific modification of K18 on Ser 30 serves as a stabilizing factor, amplifying the expression of cell cycle checkpoints. This molecular event intricately fosters cell cycle progression and augments cellular growth in CCA. Notably, the interaction between O-GlcNAcylated K18 and IDHs orchestrates metabolic reprogramming by down-regulating citrate and isocitrate levels while elevating α-ketoglutarate (α-KG). These metabolic shifts further contribute to the overall tumorigenic potential of CCA. Our study thus expands the current understanding of protein O-GlcNAcylation and introduces a new layer of complexity to post-translational control over metabolism and tumorigenesis.
Collapse
Affiliation(s)
- Xiangfeng Meng
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yue Zhou
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated, Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lei Xu
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated, Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Limu Hu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Changjiang Wang
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiao Tian
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiang Zhang
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated, Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yi Hao
- College
of
Chemistry and Molecular Engineering, Peking
University, Beijing 100871, China
| | - Bo Cheng
- School
of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jing Ma
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, China
- Collaborative
Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, China
| | - Lei Wang
- Department
of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated, Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jialin Liu
- State
Key Laboratory of Medical Proteomics, Beijing Proteome Research Center,
National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ran Xie
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry
and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
- Beijing
National Laboratory for Molecular Sciences, Beijing 100191, China
| |
Collapse
|
5
|
Sarabhai T, Kahl S, Gancheva S, Mastrototaro L, Dewidar B, Pesta D, Ratter-Rieck JM, Bobrov P, Jeruschke K, Esposito I, Schlensak M, Roden M. Loss of mitochondrial adaptation associates with deterioration of mitochondrial turnover and structure in metabolic dysfunction-associated steatotic liver disease. Metabolism 2024; 151:155762. [PMID: 38122893 DOI: 10.1016/j.metabol.2023.155762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/29/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Obesity and type 2 diabetes frequently have metabolic dysfunction-associated steatotic liver disease (MASLD) including steatohepatitis (MASH). In obesity, the liver may adapt its oxidative capacity, but the role of mitochondrial turnover in MASLD remains uncertain. METHODS This cross-sectional study compared individuals with class III obesity (n = 8/group) without (control, OBE CON; NAFLD activity score: 0.4 ± 0.1) or with steatosis (OBE MASL, 2.3 ± 0.4), or MASH (OBE MASH, 5.3 ± 0.3, p < 0.05 vs. other groups). Hepatic mitochondrial ultrastructure was assessed by transmission electron microscopy, mitochondrial respiration by high-resolution respirometry, biomarkers of mitochondrial quality control and endoplasmic reticulum (ER) stress by Western Blot. RESULTS Mitochondrial oxidative capacity was 31 % higher in OBE MASL, but 25 % lower in OBE MASH (p < 0.05 vs. OBE CON). OBE MASH showed ~1.5fold lower mitochondrial number, but ~1.2-1.5fold higher diameter and area (p < 0.001 vs. other groups). Biomarkers of autophagy (p62), mitophagy (PINK1, PARKIN), fission (DRP-1, FIS1) and fusion (MFN1/2, OPA1) were reduced in OBE MASH (p < 0.05 vs. OBE CON). OBE MASL showed lower p62, p-PARKIN/PARKIN, and p-DRP-1 (p < 0.05 vs. OBE CON). OBE MASL and MASH showed higher ER stress markers (PERK, ATF4, p-eIF2α-S51/eIF2α; p < 0.05 vs. OBE CON). Mitochondrial diameter associated inversely with fusion/fission biomarkers and with oxidative capacity, but positively with H2O2. CONCLUSION Humans with hepatic steatosis already exhibit impaired mitochondrial turnover, despite upregulated oxidative capacity, and evidence for ER stress. In MASH, oxidative stress likely mediates progressive decline of mitochondrial turnover, ultrastructure and respiration indicating that mitochondrial quality control is key for energy metabolism and may have potential for targeting MASH. ClinGovTrial:NCT01477957.
Collapse
Affiliation(s)
- Theresia Sarabhai
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sabine Kahl
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sofiya Gancheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Bedair Dewidar
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Dominik Pesta
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany; Centre for Endocrinology, Diabetes and Preventive Medicine, University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - Jacqueline M Ratter-Rieck
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
| | - Kay Jeruschke
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Irene Esposito
- Institute of Pathology, University Hospital and Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias Schlensak
- Department of Obesity and Reflux Center, Neuwerk Hospital Mönchengladbach, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany.
| |
Collapse
|
6
|
Pan L, E T, Xu C, Fan X, Xia J, Liu Y, Liu J, Zhao J, Bao N, Zhao Y, Sun H, Qin G, Farouk MH. The apoptotic effects of soybean agglutinin were induced through three different signal pathways by down-regulating cytoskeleton proteins in IPEC-J2 cells. Sci Rep 2023; 13:5753. [PMID: 37031286 PMCID: PMC10082828 DOI: 10.1038/s41598-023-32951-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/05/2023] [Indexed: 04/10/2023] Open
Abstract
Soybean agglutinin (SBA) is a main anti-nutritional factor in soybean. SBA exhibits its anti-nutritional functions by binding to intestinal epithelial cells. Keratin8 (KRT8), Keratin18 (KRT18) and Actin (ACTA) are the representative SBA-specific binding proteins. Such cytoskeletal proteins act a crucial role in different cell activities. However, limited reports reveal what the signal transduction pathway of apoptosis caused by SBA when binding to KRT8, KRT18 and ACTA. We aimed to evaluate the effects of SBA on cell apoptosis and the expression of the cytoskeletal protein (KRT8, KRT18 and ACTA), reveal the roles of these cytoskeletal proteins or their combinations on SBA-induced cell apoptosis in IPEC-J2 cell line, evaluate the influences of SBA on the mitochondria, endoplasmic reticulum stress and death receptor-mediated apoptosis signal pathway and to show the roles of KRT8, KRT18 and ACTA in different apoptosis signal pathways induced by SBA. The results showed that SBA induced the IPEC-J2 cell apoptosis and decreased the mRNA expression of KRT8, KRT18 and ACTA (p < 0.05). The degree of effect of three cytoskeleton proteins on cell apoptosis was ACTA > KRT8 > KRT18. The roles of these three cytoskeletal proteins on IPEC-J2 apoptotic rates had a certain accumulation effect. SBA up-regulated mitochondrial fission variant protein (FIS1) and fusion protein (Mfn2) promoted CytC and AIF in mitochondria to enter the cytoplasm, activated caspase-9 and caspase-3, damaged or declined mitochondrial function and reduced ATP synthesis (p < 0.05). Also, SBA up-regulated the expression of GRP78, XBP-1, eIF2α, p-eIF2α and CHOP (p < 0.05), down-regulated the expression level of ASK1 protein (p < 0.05). SBA led to the recruitment of FADD to the cytoplasmic membrane and increased the expression of FasL, resulting in caspase-8 processing. SBA up-regulated the expression level of Bax protein and decreased cytosolic Bcl-2 and Bid (p < 0.05). In addition, there was a significant negative correlation between the gene expression of cytoskeleton proteins and apoptosis, as well as the expression of key proteins of apoptosis-related signal transduction pathways. In conclusion, SBA induced the activation of the mitochondria, endoplasmic reticulum stress and the death receptor-mediated apoptosis signal pathway and the crosstalk between them. The effect of SBA on these three pathways was mainly exhibited via down-regulation of the mRNA expression of the three cytoskeletal expressions. This study elucidates the molecular mechanism and signaling pathway of SBA that lead to apoptosis from the perspective of cell biology and molecular biology and provides a new perspective on the toxicity mechanism of other food-derived anti-nutrients, medical gastrointestinal health and related cancer treatment.
Collapse
Affiliation(s)
- Li Pan
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Tianjiao E
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Chengyu Xu
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Xiapu Fan
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Jiajia Xia
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Yan Liu
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Jiawei Liu
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Jinpeng Zhao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Nan Bao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Yuan Zhao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Hui Sun
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Guixin Qin
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science, Jilin Province, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China.
| | - Mohammed Hamdy Farouk
- Animal Production Department, Faculty of Agriculture, Al-Azhar University, Nasr City, 11884, Cairo, Egypt.
| |
Collapse
|
7
|
Kim S, Mun S, Shin W, Han K, Kim MY. Identification of Potentially Pathogenic Variants Associated with Recurrence in Medication-Related Osteonecrosis of the Jaw (MRONJ) Patients Using Whole-Exome Sequencing. J Clin Med 2022; 11:jcm11082145. [PMID: 35456240 PMCID: PMC9030961 DOI: 10.3390/jcm11082145] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Bisphosphonates are antiresorptive and antiangiogenic drugs that prevent and treat bone loss and mineralization in women with postmenopausal osteoporosis and cancer patients. Medication-related osteonecrosis of the jaw (MRONJ) is commonly caused by tooth extraction and dental trauma. Although genetic and pathological studies about MRONJ have been conducted, the pathogenesis of MRONJ still remains unclear. Methods: We aimed to identify genetic variants associated with MRONJ, using whole-exome sequencing (WES). Ten MRONJ patients prescribed bisphosphonates were recruited for WES, and jawbone tissue and blood samples were collected from the patients. Results: The analysis of the WES data found a total of 1866 SNP and 40 InDel variants which are specific to MRONJ. The functional classification assay using Gene Ontology and pathway analysis discovered that genes bearing the MRONJ variants are significantly enriched for keratinization and calcium ion transport. Some of the variants are potential pathogenic variants (24 missense mutations and seven frameshift mutations) with MAF < 0.01. Conclusions: The variants are located in eight different genes (KRT18, MUC5AC, NBPF9, PABPC3, MST1L, ASPN, ATN1, and SLAIN1). Nine deleterious SNPs significantly associated with MRONJ were found in the KRT18 and PABPC3 genes. It suggests that KRT18 and PABPC3 could be MRONJ-related key genes.
Collapse
Affiliation(s)
- Songmi Kim
- Center for Bio Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea (S.M.)
- Department of Microbiology, Dankook University, Cheonan 31116, Korea
| | - Seyoung Mun
- Center for Bio Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea (S.M.)
- Department of Microbiology, Dankook University, Cheonan 31116, Korea
| | - Wonseok Shin
- NGS Clinical Laboratory, Dankook University Hospital, Cheonan 31116, Korea;
| | - Kyudong Han
- Center for Bio Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea (S.M.)
- Department of Microbiology, Dankook University, Cheonan 31116, Korea
- Correspondence: (K.H.); (M.-Y.K.); Tel.: +82-41-550-1240 (K.H.); +82-41-550-1912 (M.-Y.K.)
| | - Moon-Young Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Dankook University, Cheonan 31116, Korea
- Correspondence: (K.H.); (M.-Y.K.); Tel.: +82-41-550-1240 (K.H.); +82-41-550-1912 (M.-Y.K.)
| |
Collapse
|
8
|
Son S, Baek A, Lee JH, Kim DE. Autophagosome-lysosome fusion is facilitated by plectin-stabilized actin and keratin 8 during macroautophagic process. Cell Mol Life Sci 2022; 79:95. [PMID: 35080691 PMCID: PMC11072119 DOI: 10.1007/s00018-022-04144-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022]
Abstract
Autophagy is a lysosome-mediated degradative process that removes damaged proteins and organelles, during which autophagosome-lysosome fusion is a key step of the autophagic flux. Based on our observation that intermediate cytofilament keratin 8 (KRT8) enhances autophagic clearance in cells under oxidative stress condition, we investigated whether KRT8 supports the cytoplasmic architectural networks to facilitate the vesicular fusion entailing trafficking onto filamentous tracks. We found that KRT8 interacts with actin filaments via the cytolinker, plectin (PLEC) during trafficking of autophagosome. When PLEC was knocked down or KRT8 structure was collapsed by phosphorylation, autophagosome-lysosome fusion was attenuated. Inhibition of actin polymerization resulted in accumulation of autophagosomes owing to a decrease in autophagosome and lysosome fusion. Furthermore, myosin motor protein was found to be responsible for vesicular trafficking along the actin filaments to entail autolysosome formation. Thus, the autophagosome-lysosome fusion is aided by PLEC-stabilized actin filaments as well as intermediate cytofilament KRT8 that supports the structural integrity of actin filaments during macroautophagic process under oxidative stress condition.
Collapse
Affiliation(s)
- Sumin Son
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ahruem Baek
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Jong Hun Lee
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam, 13120, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
9
|
Shin JY, Lee J, Park J, Kim M, Chung H, Byeon SH. Association of Keratin 8 Level in Aqueous Humor With Outcomes of Intravitreal Ranibizumab Treatment for Neovascular Age-Related Macular Degeneration. Transl Vis Sci Technol 2022; 11:26. [PMID: 35040914 PMCID: PMC8764207 DOI: 10.1167/tvst.11.1.26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Purpose To investigate keratin 8 (KRT8) level in the aqueous humor (AH) of patients with neovascular age-related macular degeneration (nAMD) and elucidate its association with intravitreal ranibizumab (IVR) treatment outcomes. Methods This prospective study involved 58 eyes of treatment-naïve nAMD patients treated with three IVR doses monthly and whose AH samples were collected at baseline and two months after the initial treatment. KRT8 level was determined using the enzyme-linked immunosorbent assay and compared with that of the control group, which comprised patients who underwent cataract surgery during the same period. The nAMD-affected eyes were classified into responder (dry) and poor responder (persistent fluid) groups, according to optical coherence tomography (OCT) findings at month three. Additionally, associations between the KRT8 level and IVR treatment outcomes were analyzed. Results The baseline KRT8 level was significantly higher in the AMD group than in the control group. In the AMD group, responders demonstrated significant differences between the KRT8 level at the baseline and month two, whereas poor responders exhibited no significant change. Regression analysis revealed that a higher KRT8 level at month two was significantly associated with persistent fluid upon OCT at months three and six. Conclusions Monitoring aqueous KRT8 level may facilitate early determination of the therapeutic effects of IVR in nAMD patients and reflect the conditions of retinal pigment epithelium during the disease course. Translational Relevance Monitoring aqueous KRT8 may aid early determination of therapeutic effects of IVR in neovascular AMD patients and reflect the health conditions of retinal pigment epithelium.
Collapse
Affiliation(s)
- Joo Youn Shin
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jonghyun Lee
- Department of Ophthalmology, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Republic of Korea
| | - Jinkyu Park
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Kim
- Department of Ophthalmology, Institute of Vision Research, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyewon Chung
- Department of Ophthalmology, Konkuk University School of Medicine, Konkuk University Medical Center, Seoul, Korea
| | - Suk Ho Byeon
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Evans CA, Corfe BM. Colorectal keratins: Integrating nutrition, metabolism and colorectal health. Semin Cell Dev Biol 2021; 128:103-111. [PMID: 34481710 DOI: 10.1016/j.semcdb.2021.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 01/12/2023]
Abstract
The colon mucosa is lined with crypts of circa 300 cells, forming a continuous barrier whose roles include absorption of water, recovery of metabolic energy sources (notably short chain fatty acids), secretion of a protective mucus barrier, and physiological signalling. There is high turnover and replenishment of cells in the mucosa, disruption of this may lead to bowel pathologies including cancer and inflammatory bowel disease. Keratins have been implicated in the processes of cell death, epithelial integrity, response to inflammation and as a result are often described as guardians of the colonic epithelium. Keratin proteins carry extensive post-translational modifications, the cofactors for kinases, acetyl transferases and other modification-regulating enzymes are themselves products of metabolism. A cluster of studies has begun to reveal a bidirectional relationship between keratin form and function and metabolism. In this paper we hypothesise a mechanistic interaction between keratins and metabolism is governed through regulation of post-translational modifications and may contribute significantly to the normal functioning of the colon, placing keratins at the centre of a nutrition-metabolism-health triangle.
Collapse
Affiliation(s)
- Caroline A Evans
- ChELSI Institute, Department of Chemical and Biological Engineering, University of Sheffield, Mappin St, S1 3JD Sheffield, United Kingdom
| | - Bernard M Corfe
- Population Health Sciences Institute, Human Nutrition Research Centre, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, United Kingdom.
| |
Collapse
|
11
|
Chalar C, Clivio G, Montagne J, Costábile A, Lima A, Papa NG, Berois N, Arezo MJ. Embryonic developmental arrest in the annual killifish Austrolebias charrua: A proteomic approach to diapause III. PLoS One 2021; 16:e0251820. [PMID: 34086690 PMCID: PMC8177498 DOI: 10.1371/journal.pone.0251820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/03/2021] [Indexed: 11/18/2022] Open
Abstract
Diapause is a reversible developmental arrest faced by many organisms in harsh environments. Annual killifish present this mechanism in three possible stages of development. Killifish are freshwater teleosts from Africa and America that live in ephemeral ponds, which dry up in the dry season. The juvenile and adult populations die, and the embryos remain buried in the bottom mud until the next rainy season. Thus, species survival is entirely embryo-dependent, and they are perhaps the most remarkable extremophile organisms among vertebrates. The aim of the present study was to gather information about embryonic diapauses with the use of a "shotgun" proteomics approach in diapause III and prehatching Austrolebias charrua embryos. Our results provide insight into the molecular mechanisms of diapause III. Data are available via ProteomeXchange with identifier PXD025196. We detected a diapause-dependent change in a large group of proteins involved in different functions, such as metabolic pathways and stress tolerance, as well as proteins related to DNA repair and epigenetic modifications. Furthermore, we observed a diapause-associated switch in cytoskeletal proteins. This first glance into global protein expression differences between prehatching and diapause III could provide clues regarding the induction/maintenance of this developmental arrest in A. charrua embryos. There appears to be no single mechanism underlying diapause and the present data expand our knowledge of the molecular basis of diapause regulation. This information will be useful for future comparative approaches among different diapauses in annual killifish and/or other organisms that experience developmental arrest.
Collapse
Affiliation(s)
- Cora Chalar
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Graciela Clivio
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Jimena Montagne
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Alicia Costábile
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Analía Lima
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur Montevideo, Montevideo, Uruguay
- Instituto de Investigaciones Biológicas Clemente Estable, Ministerio de Educación y Cultura, Montevideo, Uruguay
| | - Nicolás G. Papa
- Laboratorio de Biología Molecular de Organismos Acuáticos, Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Nibia Berois
- Laboratorio de Biología Molecular de Organismos Acuáticos, Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María José Arezo
- Laboratorio de Biología Molecular de Organismos Acuáticos, Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- * E-mail:
| |
Collapse
|
12
|
Molecular Tumor Subtypes of HPV-Positive Head and Neck Cancers: Biological Characteristics and Implications for Clinical Outcomes. Cancers (Basel) 2021; 13:cancers13112721. [PMID: 34072836 PMCID: PMC8198180 DOI: 10.3390/cancers13112721] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 01/18/2023] Open
Abstract
Until recently, research on the molecular signatures of Human papillomavirus (HPV)-associated head and neck cancers mainly focused on their differences with respect to HPV-negative head and neck squamous cell carcinomas (HNSCCs). However, given the continuing high incidence level of HPV-related HNSCC, the time is ripe to characterize the heterogeneity that exists within these cancers. Here, we review research thus far on HPV-positive HNSCC molecular subtypes, and their relationship with clinical characteristics and HPV integration into the host genome. Different omics data including host transcriptomics and epigenomics, as well as HPV characteristics, can provide complementary viewpoints. Keratinization, mesenchymal differentiation, immune signatures, stromal cells and oxidoreductive processes all play important roles.
Collapse
|
13
|
Baek A, Son S, Baek YM, Kim DE. KRT8 (keratin 8) attenuates necrotic cell death by facilitating mitochondrial fission-mediated mitophagy through interaction with PLEC (plectin). Autophagy 2021; 17:3939-3956. [PMID: 33783309 DOI: 10.1080/15548627.2021.1897962] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Dysregulation of mitochondrial homeostasis and accumulation of damaged mitochondria cause degenerative diseases such as age-related macular degeneration (AMD). We studied the effects of the intermediate cytofilament KRT8 (keratin 8) on mitochondrial homeostasis in relation to the morphology and function of mitochondria in retinal pigment epithelial cells under oxidative stress. When the mitochondria were damaged owing to oxidative stress, the damaged mitochondria were readily disposed of via mitophagy following mitochondrial fission. During this process, KRT8 was found to physically interact with the mitochondria through PLEC (plectin) and facilitate the mitochondrial fission-mediated mitophagy. However, the association between PLEC-anchoring mitochondria and KRT8 was dwindled by KRT8 phosphorylation under oxidative stress. The efficient KRT8-facilitated mitophagy flux suppressed the accumulation of damaged mitochondria and consequently diminished necrotic cell death under oxidative stress. Thus, by facilitating mitophagy, KRT8 protects RPE cells against necrotic cell death due to oxidative stress.
Collapse
Affiliation(s)
- Ahruem Baek
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Republic of Korea
| | - Sumin Son
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Republic of Korea
| | - Yu Mi Baek
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Republic of Korea
| |
Collapse
|
14
|
Yin S, Shi Q, Shao W, Zhang C, Zhang Y, Qiu X, Huang J. Hepatocyte-Derived Igκ Exerts a Protective Effect against ConA-Induced Acute Liver Injury. Int J Mol Sci 2020; 21:ijms21249379. [PMID: 33317072 PMCID: PMC7763521 DOI: 10.3390/ijms21249379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/02/2022] Open
Abstract
Immunoglobulin (Igκ) has been reported to be expressed in sorted liver epithelial cells of μMT mice, and the sequence characteristics of hepatocyte-derived Igκ were different from those of classical B-cell-derived Igκ. However, the physiological function of hepatocyte-derived Igκ is still unclear. The expression of Igκ was firstly identified in primary hepatocytes and normal liver cell line (NCTC1469), and hepatocyte-derived Igκ expression was elevated and displayed unique localization in hepatocytes of concanavalin A (ConA)-induced hepatitis model. Moreover, Igκ knockout mice were more sensitive to ConA-induced hepatitis and had higher serum aspartate aminotransferase (AST) levels, more severe histological injury and a greater number of terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL)-positive cells as compared with littermate controls. Furthermore, knockdown of Igκ in primary hepatocytes and NCTC1469 cells led to accelerated activation of the mitochondrial death pathway and caspase-3 cleavage in vitro, which might be related to inhibition of NF-κB signaling pathway and activation of JNK via the cytoskeleton dynamics. Taken together, these results indicate that hepatocyte-derived Igκ mediates cellular resistance to ConA-induced liver injury by inhibiting activation of caspase-3 and the mitochondrial death pathway, suggesting that Igκ plays an important role in hepatocyte survival and exerts a protective effect against ConA-induced liver injury in mice.
Collapse
Affiliation(s)
- Sha Yin
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Qianwen Shi
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Wenwei Shao
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Chi Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Yixiao Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
- Correspondence: (X.Q.); (J.H.); Tel.: +86-10-82805744 (X.Q.); +86-10-82802846 (J.H.)
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (S.Y.); (Q.S.); (W.S.); (C.Z.); (Y.Z.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
- Correspondence: (X.Q.); (J.H.); Tel.: +86-10-82805744 (X.Q.); +86-10-82802846 (J.H.)
| |
Collapse
|
15
|
Steen K, Chen D, Wang F, Majumdar R, Chen S, Kumar S, Lombard DB, Weigert R, Zieman AG, Parent CA, Coulombe PA. A role for keratins in supporting mitochondrial organization and function in skin keratinocytes. Mol Biol Cell 2020; 31:1103-1111. [PMID: 32213122 PMCID: PMC7353162 DOI: 10.1091/mbc.e19-10-0565] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondria fulfill essential roles in ATP production, metabolic regulation, calcium signaling, generation of reactive oxygen species (ROS), and additional determinants of cellular health. Recent studies have highlighted a role for mitochondria during cell differentiation, including in skin epidermis. The observation of oxidative stress in keratinocytes from Krt16 null mouse skin, a model for pachyonychia congenita (PC)–associated palmoplantar keratoderma, prompted us to examine the role of Keratin (K) 16 protein and its partner K6 in regulating the structure and function of mitochondria. Electron microscopy revealed major anomalies in mitochondrial ultrastructure in late stage, E18.5, Krt6a/Krt6b null embryonic mouse skin. Follow-up studies utilizing biochemical, metabolic, and live imaging readouts showed that, relative to controls, skin keratinocytes null for Krt6a/Krt6b or Krt16 exhibit elevated ROS, reduced mitochondrial respiration, intracellular distribution differences, and altered movement of mitochondria within the cell. These findings highlight a novel role for K6 and K16 in regulating mitochondrial morphology, dynamics, and function and shed new light on the causes of oxidative stress observed in PC and related keratin-based skin disorders.
Collapse
Affiliation(s)
- Kaylee Steen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Desu Chen
- Laboratory for Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Fengrong Wang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Ritankar Majumdar
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Song Chen
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Surinder Kumar
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - David B Lombard
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Roberto Weigert
- Laboratory for Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Abigail G Zieman
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Carole A Parent
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Pierre A Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
16
|
Phosphorylation of keratin 18 serine 52 regulates mother-daughter centriole engagement and microtubule nucleation by cell cycle-dependent accumulation at the centriole. Histochem Cell Biol 2020; 153:307-321. [PMID: 32078038 DOI: 10.1007/s00418-020-01849-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
Serine-52 (Ser52) is the major physiologic site of keratin 18 (K18) phosphorylation. Here, we report that serine-52 phosphorylated K18 (phospho-Ser52 K18) accumulated on centrosomes in a cell cycle-dependent manner. Moreover, we found that phospho-Ser52 K18 was located at the proximal end of the mother centriole. Transfection with the K18 Ser52 → Ala (K18 S52A) mutant prevented centriole localization of phospho-Ser52 K18 and resulted in separation of the mother-daughter centrioles. Inhibition of microtubule polymerization led to the disappearance of aggregated phospho-Ser52 K18 on the centrosome; removal of inhibitors resulted in reaccumulation of phospho-Ser52 K18 in microtubule-organizing centers. Transfection with a K18 S52A mutant inhibited microtubule nucleation. These results reveal a cell cycle-dependent change in centrosome localization of phospho-Ser52 k18 and strongly suggest that the phosphorylation status of Ser52 K18 of mother centrioles plays a critical role in maintaining a tight engagement between mother and daughter centrioles and also contributes to microtubule nucleation.
Collapse
|
17
|
Miao Q, Xu Y, Yin H, Zhang H, Ye J. KRT8 phosphorylation regulates the epithelial-mesenchymal transition in retinal pigment epithelial cells through autophagy modulation. J Cell Mol Med 2020; 24:3217-3228. [PMID: 32022439 PMCID: PMC7077598 DOI: 10.1111/jcmm.14998] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/15/2019] [Accepted: 12/24/2019] [Indexed: 01/13/2023] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a severe ocular disease which results in complex retinal detachment and irreversible vision loss. The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is considered to be critical in the pathogenesis of PVR. In this study, we focused on the potential impact of keratin 8 (KRT8) phosphorylation and autophagy on TGF-β2-induced EMT of RPE cells and explored the relationship between them. Using immunofluorescence and Western blot analysis, the co-localization of KRT8 and autophagy marker, as well as the abundance of phosphorylated KRT8 (p-KRT8) expression, was observed within subretinal and epiretinal membranes from PVR patients. Moreover, during TGF-β2-induced EMT process, we found that p-KRT8 was enhanced in RPE cells, which accompanied by an increase in autophagic flux. Inhibition of autophagy with pharmacological inhibitors or specific siRNAs was associated with a reduction in cell migration and the synthesis of several EMT markers. In the meantime, we demonstrated that p-KRT8 was correlated with the autophagy progression during the EMT of RPE cells. Knockdown the expression or mutagenesis of the critical phosphorylated site of KRT8 would induce autophagy impairment, through affecting the fusion of autophagosomes and lysosomes. Therefore, this study may provide a new insight into the pathogenesis of PVR and suggests the potential therapeutic value of p-KRT8 in the prevention and treatment of PVR.
Collapse
Affiliation(s)
- Qi Miao
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yufeng Xu
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Houfa Yin
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huina Zhang
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Ye
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Abo El-khair SM, Ghoneim FM, Shabaan DA, Elsamanoudy AZ. Molecular and ultrastructure study of endoplasmic reticulum stress in hepatic steatosis: role of hepatocyte nuclear factor 4α and inflammatory mediators. Histochem Cell Biol 2019; 153:49-62. [PMID: 31637472 DOI: 10.1007/s00418-019-01823-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
|
19
|
Klymkowsky MW. Filaments and phenotypes: cellular roles and orphan effects associated with mutations in cytoplasmic intermediate filament proteins. F1000Res 2019; 8. [PMID: 31602295 PMCID: PMC6774051 DOI: 10.12688/f1000research.19950.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic intermediate filaments (IFs) surround the nucleus and are often anchored at membrane sites to form effectively transcellular networks. Mutations in IF proteins (IFps) have revealed mechanical roles in epidermis, muscle, liver, and neurons. At the same time, there have been phenotypic surprises, illustrated by the ability to generate viable and fertile mice null for a number of IFp-encoding genes, including vimentin. Yet in humans, the vimentin ( VIM) gene displays a high probability of intolerance to loss-of-function mutations, indicating an essential role. A number of subtle and not so subtle IF-associated phenotypes have been identified, often linked to mechanical or metabolic stresses, some of which have been found to be ameliorated by the over-expression of molecular chaperones, suggesting that such phenotypes arise from what might be termed "orphan" effects as opposed to the absence of the IF network per se, an idea originally suggested by Toivola et al. and Pekny and Lane.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
20
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
21
|
Animal models of NAFLD from the pathologist's point of view. Biochim Biophys Acta Mol Basis Dis 2018; 1865:929-942. [PMID: 29746920 DOI: 10.1016/j.bbadis.2018.04.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 01/18/2023]
Abstract
Fatty liver disease is a multifactorial world-wide health problem resulting from a complex interplay between liver, adipose tissue and intestine and initiated by alcohol abuse, overeating, various types of intoxication, adverse drug reactions and genetic or acquired metabolic defects. Depending on etiology fatty liver disease is commonly categorized as alcoholic or non-alcoholic. Both types may progress from simple steatosis to the necro-inflammatory lesion of alcoholic (ASH) and non-alcoholic steatohepatitis (NASH), respectively, and finally to cirrhosis and hepatocellular carcinoma. Animal models are helpful to clarify aspects of pathogenesis and progression. Generally, they are classified as nutritional (dietary), toxin-induced and genetic, respectively, or represent a combination of these factors. Numerous reviews are dealing with NASH animal models designed to imitate as closely as possible the metabolic situation associated with human disease. This review focuses on currently used mouse models of NASH with particular emphasis on liver morphology. Despite metabolic similarities most models (except those with chemically or genetically induced porphyria or keratin 18-deficiency) fail to develop the morphologic key features of NASH, namely hepatocyte ballooning and formation of histologically and immunohistochemically well-defined Mallory-Denk-Bodies (MDBs). Although MDBs are not universally detectable in ballooned hepatocytes in NASH their experimental reproduction and analysis may, however, significantly contribute to our understanding of important pathogenic aspects of NASH despite the obvious differences in etiology.
Collapse
|
22
|
Stanley RL, Ohashi T, Gordon J, Mowa CN. A proteomic profile of postpartum cervical repair in mice. J Mol Endocrinol 2018; 60:17-28. [PMID: 29259042 DOI: 10.1530/jme-17-0179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/15/2017] [Indexed: 01/01/2023]
Abstract
A timely and complete uterine cervical tissue repair postpartum is of necessity to prevent obstetrical complications, such as cervicitis, ectropion, hemorrhage, repeated miscarriages or abortions and possibly preterm labor and malignancies. We recently characterized the morphological alterations, as well as changes in angiogenic expression profile in a mice uterine cervix during the immediate postpartum period. Here, we build on this previous study using a proteomic analysis to profile postpartum tissue changes in mice cervix during the same period, the first 48 h of postpartum. The current proteomics data reveal a variable expression of several intermediate filaments, cytoskeletal modulators and proteins with immune and/or wound-healing properties. We conclude that postpartum cervical repair involves a rapid and tightly regulated balance between a host of biological factors, notably between anti- and pro-inflammatory factors, executed by the M1 and M2 macrophage cells, as revealed by proteomics and verified by confocal immunofluorescence. Future studies will assess the suitability of some of the key proteins identified in this study as potential markers for determining the phase of postpartum cervical repair in obstetrical complications, such as cervical lacerations.
Collapse
Affiliation(s)
- Robert Lee Stanley
- Department of BiologyAppalachian State University, Boone, North Carolina, USA
| | - Takako Ohashi
- Department of BiologyAppalachian State University, Boone, North Carolina, USA
| | - Jacob Gordon
- Department of BiologyAppalachian State University, Boone, North Carolina, USA
| | | |
Collapse
|
23
|
Effects of aspirin and clopidogrel on neural stem cells. Cell Biol Toxicol 2017; 34:219-232. [DOI: 10.1007/s10565-017-9412-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022]
|
24
|
Omary MB. Intermediate filament proteins of digestive organs: physiology and pathophysiology. Am J Physiol Gastrointest Liver Physiol 2017; 312:G628-G634. [PMID: 28360031 PMCID: PMC5495917 DOI: 10.1152/ajpgi.00455.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 01/31/2023]
Abstract
Intermediate filament proteins (IFs), such as cytoplasmic keratins in epithelial cells and vimentin in mesenchymal cells and the nuclear lamins, make up one of the three major cytoskeletal protein families. Whether in digestive organs or other tissues, IFs share several unique features including stress-inducible overexpression, abundance, cell-selective and differentiation state expression, and association with >80 human diseases when mutated. Whereas most IF mutations cause disease, mutations in simple epithelial keratins 8, 18, or 19 or in lamin A/C predispose to liver disease with or without other tissue manifestations. Keratins serve major functions including protection from apoptosis, providing cellular and subcellular mechanical integrity, protein targeting to subcellular compartments, and scaffolding and regulation of cell-signaling processes. Keratins are essential for Mallory-Denk body aggregate formation that occurs in association with several liver diseases, whereas an alternate type of keratin and lamin aggregation occurs upon liver involvement in porphyria. IF-associated diseases have no known directed therapy, but high-throughput drug screening to identify potential therapies is an appealing ongoing approach. Despite the extensive current knowledge base, much remains to be discovered regarding IF physiology and pathophysiology in digestive and nondigestive organs.
Collapse
Affiliation(s)
- M. Bishr Omary
- Department of Molecular and Integrative Physiology and Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
25
|
Baek A, Yoon S, Kim J, Baek YM, Park H, Lim D, Chung H, Kim DE. Autophagy and KRT8/keratin 8 protect degeneration of retinal pigment epithelium under oxidative stress. Autophagy 2017; 13:248-263. [PMID: 28045574 DOI: 10.1080/15548627.2016.1256932] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Contribution of autophagy and regulation of related proteins to the degeneration of retinal pigment epithelium (RPE) in age-related macular degeneration (AMD) remain unknown. We report that upregulation of KRT8 (keratin 8) as well as its phosphorylation are accompanied with autophagy and attenuated with the inhibition of autophagy in RPE cells under oxidative stress. KRT8 appears to have a dual role in RPE pathophysiology. While increased expression of KRT8 following autophagy provides a cytoprotective role in RPE, phosphorylation of KRT8 induces pathologic epithelial-mesenchymal transition (EMT) of RPE cells under oxidative stress, which is mediated by MAPK1/ERK2 (mitogen-activated protein kinase 1) and MAPK3/ERK1. Inhibition of autophagy further promotes EMT, which can be reversed by inhibition of MAPK. Thus, regulated enhancement of autophagy with concurrent increased expression of KRT8 and the inhibition of KRT8 phosphorylation serve to inhibit oxidative stress-induced EMT of RPE cells as well as to prevent cell death, suggesting that pharmacological manipulation of KRT8 upregulation through autophagy with combined inhibition of the MAPK1/3 pathway may be attractive therapeutic strategies for the treatment of AMD.
Collapse
Affiliation(s)
- Ahruem Baek
- a Department of Bioscience and Biotechnology , Konkuk University , Gwangjin-gu , Seoul , Korea
| | - Soojin Yoon
- a Department of Bioscience and Biotechnology , Konkuk University , Gwangjin-gu , Seoul , Korea
| | - Jean Kim
- b Department of Ophthalmology , Konkuk University School of Medicine , Gwangjin-gu , Seoul , Korea
| | - Yu Mi Baek
- a Department of Bioscience and Biotechnology , Konkuk University , Gwangjin-gu , Seoul , Korea
| | - Hanna Park
- a Department of Bioscience and Biotechnology , Konkuk University , Gwangjin-gu , Seoul , Korea
| | - Daehan Lim
- b Department of Ophthalmology , Konkuk University School of Medicine , Gwangjin-gu , Seoul , Korea
| | - Hyewon Chung
- b Department of Ophthalmology , Konkuk University School of Medicine , Gwangjin-gu , Seoul , Korea
| | - Dong-Eun Kim
- a Department of Bioscience and Biotechnology , Konkuk University , Gwangjin-gu , Seoul , Korea
| |
Collapse
|
26
|
|
27
|
Ku NO, Strnad P, Bantel H, Omary MB. Keratins: Biomarkers and modulators of apoptotic and necrotic cell death in the liver. Hepatology 2016; 64:966-76. [PMID: 26853542 PMCID: PMC4977204 DOI: 10.1002/hep.28493] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Keratins, formerly known as cytokeratins, are the major epithelial-specific subgroup of intermediate filament proteins. Adult hepatocytes express keratin polypeptides 8 and 18 (K8/K18), whereas cholangiocytes express K8/K18 and keratins 7 and 19 (K7/K19). Keratins function primarily to protect hepatocytes from apoptosis and necrosis, which was revealed using several genetic mouse models. This cytoprotective function was further clarified by the identification of natural human keratin variants that are normally silent, but become pathogenic by predisposing their carriers to apoptosis during acute or chronic liver injury mediated by toxins, virus infection, or metabolic stress. During apoptosis, caspases cleave K18 and K19 at conserved aspartates (human K18/K19: (235) Val-Glu-Val-Asp(↓) ) and K18 at a unique aspartate (human K18: (394) Asp-Ala-Leu-Asp(↓) ), with the latter exposed epitope becoming recognized by the M30 antibody in blood and tissues. Additional K18-containing protein backbone epitopes are detected using the M6 and M5 (termed M65) antibodies. Intact K18 and its associated fragments, which are released into blood during apoptosis and necrosis in various diseases, have been analyzed by enzyme-linked immunosorbent assay using the M30/M65 antibodies or their signal ratios. Furthermore, M30/M65 levels have been used as diagnostic and prognostic biomarkers in acute and chronic liver diseases, including nonalcoholic steatohepatitis and acute liver failure. Other keratin biomarkers include K8/K18/K19-related tissue polypeptide antigen, K18-related tissue polypeptide-specific antigen, and K19-related CYFRA-21-1, which have been evaluated mostly in patients with epithelial tumors. CONCLUSION Keratins and their fragments are released into blood during liver and other epithelial tissue injury. The epithelial specificity of K18/K19, epitope unmasking upon caspase digestion, keratin abundance, and relative keratin stability render them useful biomarkers for hepatocyte and cholangiocyte apoptosis and necrosis. However, the precise biochemical nature and release mechanism of circulating keratins remain unknown. (Hepatology 2016;64:966-976).
Collapse
Affiliation(s)
- Nam-On Ku
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul 120-749, Korea
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - M. Bishr Omary
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 7744 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622,Department of Medicine, University of Michigan Medical School, 7744 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622; and VA Ann Arbor Health Care System, Ann Arbor MI, 48105
| |
Collapse
|
28
|
Intermediate Filaments as Organizers of Cellular Space: How They Affect Mitochondrial Structure and Function. Cells 2016; 5:cells5030030. [PMID: 27399781 PMCID: PMC5040972 DOI: 10.3390/cells5030030] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/24/2016] [Accepted: 06/30/2016] [Indexed: 12/17/2022] Open
Abstract
Intermediate filaments together with actin filaments and microtubules form the cytoskeleton, which is a complex and highly dynamic 3D network. Intermediate filaments are the major mechanical stress protectors but also affect cell growth, differentiation, signal transduction, and migration. Using intermediate filament-mitochondrial crosstalk as a prominent example, this review emphasizes the importance of intermediate filaments as crucial organizers of cytoplasmic space to support these functions. We summarize observations in different mammalian cell types which demonstrate how intermediate filaments influence mitochondrial morphology, subcellular localization, and function through direct and indirect interactions and how perturbations of these interactions may lead to human diseases.
Collapse
|
29
|
Kakade PS, Budnar S, Kalraiya RD, Vaidya MM. Functional Implications of O-GlcNAcylation-dependent Phosphorylation at a Proximal Site on Keratin 18. J Biol Chem 2016; 291:12003-13. [PMID: 27059955 DOI: 10.1074/jbc.m116.728717] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 01/16/2023] Open
Abstract
Keratins 8/18 (K8/18) are phosphoglycoproteins and form the major intermediate filament network of simple epithelia. The three O-GlcNAcylation (Ser(29), Ser(30), and Ser(48)) and two phosphorylation (Ser(33) and Ser(52)) serine sites on K18 are well characterized. Both of these modifications have been reported to increase K18 solubility and regulate its filament organization. In this report, we investigated the site-specific interplay between these two modifications in regulating the functional properties of K18, like solubility, stability, and filament organization. An immortalized hepatocyte cell line (HHL-17) stably expressing site-specific single, double, and triple O-GlcNAc and phosphomutants of K18 were used to identify the site(s) critical for regulating these functions. Keratin 18 mutants where O-GlcNAcylation at Ser(30) was abolished (K18-S30A) exhibited reduced phosphorylation induced solubility, increased stability, defective filament architecture, and slower migration. Interestingly, K18-S30A mutants also showed loss of phosphorylation at Ser(33), a modification known to regulate the solubility of K18. Further to this, the K18 phosphomutant (K18-S33A) mimicked K18-S30A in its stability, filament organization, and cell migration. These results indicate that O-GlcNAcylation at Ser(30) promotes phosphorylation at Ser(33) to regulate the functional properties of K18 and also impact cellular processes like migration. O-GlcNAcylation and phosphorylation on the same or adjacent sites on most proteins antagonize each other in regulating protein functions. Here we report a novel, positive interplay between O-GlcNAcylation and phosphorylation at adjacent sites on K18 to regulate its fundamental properties.
Collapse
Affiliation(s)
- Poonam S Kakade
- From the Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India
| | - Srikanth Budnar
- From the Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India
| | - Rajiv D Kalraiya
- From the Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India
| | - Milind M Vaidya
- From the Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC), Tata Memorial Centre, Sector 22, Kharghar, Navi Mumbai 410210, India
| |
Collapse
|
30
|
Kumar V, Bouameur JE, Bär J, Rice RH, Hornig-Do HT, Roop DR, Schwarz N, Brodesser S, Thiering S, Leube RE, Wiesner RJ, Vijayaraj P, Brazel CB, Heller S, Binder H, Löffler-Wirth H, Seibel P, Magin TM. A keratin scaffold regulates epidermal barrier formation, mitochondrial lipid composition, and activity. J Cell Biol 2016; 211:1057-75. [PMID: 26644517 PMCID: PMC4674273 DOI: 10.1083/jcb.201404147] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epidermal keratin filaments are important components and organizers of the cornified envelope and regulate mitochondrial metabolism by modulating their membrane composition. Keratin intermediate filaments (KIFs) protect the epidermis against mechanical force, support strong adhesion, help barrier formation, and regulate growth. The mechanisms by which type I and II keratins contribute to these functions remain incompletely understood. Here, we report that mice lacking all type I or type II keratins display severe barrier defects and fragile skin, leading to perinatal mortality with full penetrance. Comparative proteomics of cornified envelopes (CEs) from prenatal KtyI−/− and KtyII−/−K8 mice demonstrates that absence of KIF causes dysregulation of many CE constituents, including downregulation of desmoglein 1. Despite persistence of loricrin expression and upregulation of many Nrf2 targets, including CE components Sprr2d and Sprr2h, extensive barrier defects persist, identifying keratins as essential CE scaffolds. Furthermore, we show that KIFs control mitochondrial lipid composition and activity in a cell-intrinsic manner. Therefore, our study explains the complexity of keratinopathies accompanied by barrier disorders by linking keratin scaffolds to mitochondria, adhesion, and CE formation.
Collapse
Affiliation(s)
- Vinod Kumar
- Translational Centre for Regenerative Medicine Leipzig, University of Leipzig, 04103 Leipzig, Germany Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Jamal-Eddine Bouameur
- Translational Centre for Regenerative Medicine Leipzig, University of Leipzig, 04103 Leipzig, Germany Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Janina Bär
- Translational Centre for Regenerative Medicine Leipzig, University of Leipzig, 04103 Leipzig, Germany Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Robert H Rice
- Department of Environmental Toxicology, University of California, Davis, Davis, CA 95616
| | - Hue-Tran Hornig-Do
- Center for Physiology and Pathophysiology, Institute for Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Dennis R Roop
- Department of Dermatology, University of Colorado, Denver, CO 80045 Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Denver, CO 80045
| | - Nicole Schwarz
- Institute of Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany
| | - Susanne Brodesser
- Center for Physiology and Pathophysiology, Institute for Vegetative Physiology, University of Cologne, 50931 Cologne, Germany Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Medical Faculty, University of Cologne, 50931 Cologne, Germany Center for Molecular Medicine Cologne, 50931 Cologne, Germany
| | - Sören Thiering
- Translational Centre for Regenerative Medicine Leipzig, University of Leipzig, 04103 Leipzig, Germany Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074 Aachen, Germany
| | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute for Vegetative Physiology, University of Cologne, 50931 Cologne, Germany Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Medical Faculty, University of Cologne, 50931 Cologne, Germany Center for Molecular Medicine Cologne, 50931 Cologne, Germany
| | | | - Christina B Brazel
- Translational Centre for Regenerative Medicine Leipzig, University of Leipzig, 04103 Leipzig, Germany Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Sandra Heller
- Center for Biotechnology and Biomedicine, 04103 Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany
| | - Henry Löffler-Wirth
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany
| | - Peter Seibel
- Center for Biotechnology and Biomedicine, 04103 Leipzig, Germany
| | - Thomas M Magin
- Translational Centre for Regenerative Medicine Leipzig, University of Leipzig, 04103 Leipzig, Germany Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
31
|
Roux A, Gilbert S, Loranger A, Marceau N. Impact of keratin intermediate filaments on insulin-mediated glucose metabolism regulation in the liver and disease association. FASEB J 2015; 30:491-502. [PMID: 26467793 DOI: 10.1096/fj.15-277905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/21/2015] [Indexed: 12/17/2022]
Abstract
In all cells, a tight regulation exists between glucose uptake and utilization to prevent diseases related to its perturbed metabolism. In insulin-targeted cells, such as hepatocytes, proper glucose utilization requires an elaborate interplay between the insulin receptor, the glucose transporter, and mitochondria that involves the participation of actin microfilaments and microtubules. In addition, there is increasing evidence of an involvement of the third cytoskeletal network provided by intermediate filaments (IFs). Keratins belong to the multigene family of IF proteins, coordinately expressed as distinct pairs within the context of epithelial cell differentiation. Hepatocyte IFs are made up of the [keratin (K)8/K18] pair only, whereas pancreatic β-cell IFs additionally include small amounts of K7. There are accumulating examples of K8/K18 involvement in the glucose-insulin cross-talk, including the modulation of plasma glucose levels, insulin release from pancreatic β-cells, and insulin-mediated glucose uptake and glycogen production in hepatocytes after a K8/K18 loss. This review integrates the mechanistic features that support such an impact of K8/K18 IFs on insulin-dependent glucose metabolism regulation in liver and its implication in glucose- or insulin-associated diseases.
Collapse
Affiliation(s)
- Alexandra Roux
- Centre de Recherche sur le Cancer, Université Laval, and Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada
| | - Stéphane Gilbert
- Centre de Recherche sur le Cancer, Université Laval, and Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada
| | - Anne Loranger
- Centre de Recherche sur le Cancer, Université Laval, and Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada
| | - Normand Marceau
- Centre de Recherche sur le Cancer, Université Laval, and Centre de Recherche du Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada
| |
Collapse
|
32
|
Wang J, Cai J, Huang Y, Ke Q, Wu B, Wang S, Han X, Wang T, Wang Y, Li W, Lao C, Song W, Xiang AP. Nestin regulates proliferation and invasion of gastrointestinal stromal tumor cells by altering mitochondrial dynamics. Oncogene 2015; 35:3139-50. [PMID: 26434586 DOI: 10.1038/onc.2015.370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 08/21/2015] [Accepted: 08/28/2015] [Indexed: 12/11/2022]
Abstract
Nestin is widely expressed in numerous tumors and has become a diagnostic and prognostic indicator. However, the exact mechanism by which nestin contributes to tumor malignancy remains poorly understood. Here, we found marked upregulation of nestin expression in highly proliferative and invasive gastrointestinal stromal tumor (GIST) specimens. Nestin knockdown in GIST cells reduced the proliferative and invasive activity owing to a decrease of mitochondrial intracellular reactive oxygen species (ROS) generation. Furthermore, nestin was co-localized with mitochondria, and knockdown of nestin increased mitochondrial elongation and influenced the mitochondrial function, including oxygen consumption rates, ATP generation and mitochondrial membrane potential and so on. In exploring the underlying mechanism, we demonstrated nestin knockdown inhibited the mitochondrial recruitment of Dynamin-related protein1 and induced the change of mitochondrial dynamics. Thus, nestin may have an important role in GIST malignancy by regulating mitochondrial dynamics and altering intracellular ROS levels. The findings provide new clues to reveal mechanisms by which nestin mediates the proliferation and invasion of GISTs.
Collapse
Affiliation(s)
- J Wang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - J Cai
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Biotherapy Center, Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Y Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Q Ke
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Department of Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - B Wu
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - S Wang
- Department of Gastrointestinal-Pancreatic Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - X Han
- Department of Population Genetics and Prevention, Fuwai Hospital of Peking Union Medical College, Beijing, China
| | - T Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Y Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - W Li
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - C Lao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - W Song
- Department of Gastrointestinal-Pancreatic Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - A P Xiang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Biotherapy Center, Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
33
|
Toivola DM, Habtezion A, Misiorek JO, Zhang L, Nyström JH, Sharpe O, Robinson WH, Kwan R, Omary MB. Absence of keratin 8 or 18 promotes antimitochondrial autoantibody formation in aging male mice. FASEB J 2015; 29:5081-9. [PMID: 26399787 DOI: 10.1096/fj.14-269795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 09/08/2015] [Indexed: 12/16/2022]
Abstract
Human mutations in keratin 8 (K8) and keratin 18 (K18), the intermediate filament proteins of hepatocytes, predispose to several liver diseases. K8-null mice develop chronic liver injury and fragile hepatocytes, dysfunctional mitochondria, and Th2-type colitis. We tested the hypothesis that autoantibody formation accompanies the liver damage that associates with K8/K18 absence. Sera from wild-type control, K8-null, and K18-null mice were analyzed by immunoblotting and immunofluorescence staining of cell and mouse tissue homogenates. Autoantibodies to several antigens were identified in 81% of K8-null male mice 8 mo or older. Similar autoantibodies were detected in aging K18-null male mice that had a related liver phenotype but normal colon compared with K8-null mice, suggesting that the autoantibodies are linked to liver rather than colonic disease. However, these autoantibodies were not observed in nontransgenic mice subjected to 4 chronic injury models. The autoantigens are ubiquitous and partition with mitochondria. Mass spectrometry and purified protein analysis identified, mitochondrial HMG-CoA synthase, aldehyde dehydrogenase, and catalase as the primary autoantigens, and glutamate dehydrogenase and epoxide hydrolase-2 as additional autoantigens. Therefore, absence of the hepatocyte keratins results in production of anti-mitochondrial autoantibodies (AMA) that recognize proteins involved in energy metabolism and oxidative stress, raising the possibility that AMA may be found in patients with keratin mutations that associate with liver and other diseases.
Collapse
Affiliation(s)
- Diana M Toivola
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Aida Habtezion
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Julia O Misiorek
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Linxing Zhang
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Joel H Nyström
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Orr Sharpe
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - William H Robinson
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - Raymond Kwan
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| | - M Bishr Omary
- *Department of Science and Engineering, Department of Biosciences, and Department of Cell Biology, Åbo Akademi University, Turku, Finland; Division of Gastroenterology and Hepatology, Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; and Veterans Affairs Ann Arbor Health Care System, Ann Arbor, Michigan, USA
| |
Collapse
|
34
|
Guldiken N, Zhou Q, Kucukoglu O, Rehm M, Levada K, Gross A, Kwan R, James LP, Trautwein C, Omary MB, Strnad P. Human keratin 8 variants promote mouse acetaminophen hepatotoxicity coupled with c-jun amino-terminal kinase activation and protein adduct formation. Hepatology 2015; 62:876-86. [PMID: 25963979 PMCID: PMC4549164 DOI: 10.1002/hep.27891] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 04/15/2015] [Accepted: 05/03/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED Keratins 8 and 18 (K8/K18) are the intermediate filaments proteins of simple-type digestive epithelia and provide important cytoprotective function. K8/K18 variants predispose humans to chronic liver disease progression and poor outcomes in acute acetaminophen (APAP)-related liver failure. Given that K8 G62C and R341H/R341C are common K8 variants in European and North American populations, we studied their biological significance using transgenic mice. Mice that overexpress the human K8 variants, R341H or R341C, were generated and used together with previously described mice that overexpress wild-type K8 or K8 G62C. Mice were injected with 600 mg/kg of APAP or underwent bile duct ligation (BDL). Livers were evaluated by microarray analysis, quantitative real-time polymerase chain reaction, immunoblotting, histological and immunological staining, and biochemical assays. Under basal conditions, the K8 G62C/R341H/R341C variant-expressing mice did not show an obvious liver phenotype or altered keratin filament distribution, whereas K8 G62C/R341C animals had aberrant disulphide cross-linked keratins. Animals carrying the K8 variants displayed limited gene expression changes, but had lower nicotinamide N-methyl transferase (NNMT) levels and were predisposed to APAP-induced hepatotoxicity. NNMT represents a novel K8/K18-associated protein that becomes up-regulated after K8/K18 transfection. The more pronounced liver damage was accompanied by increased and prolonged JNK activation; elevated APAP protein adducts; K8 hyperphosphorylation at S74/S432 with enhanced keratin solubility; and prominent pericentral keratin network disruption. No differences in APAP serum levels, glutathione, or adenosine triphosphate levels were noted. BDL resulted in similar liver injury and biliary fibrosis in all mouse genotypes. CONCLUSION Expression of human K8 variants G62C, R341H, or R341C in mice predisposes to acute APAP hepatotoxicity, thereby providing direct evidence for the importance of these variants in human acute liver failure.
Collapse
Affiliation(s)
- Nurdan Guldiken
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany,Department of Internal Medicine I, University Hospital Ulm, Ulm Germany
| | - Qin Zhou
- Department of Medicine, Palo Alto VA Medical Center, CA; and Stanford University Digestive Disease Center, USA
| | - Ozlem Kucukoglu
- Department of Internal Medicine I, University Hospital Ulm, Ulm Germany
| | - Melanie Rehm
- Department of Internal Medicine I, University Hospital Ulm, Ulm Germany
| | - Kateryna Levada
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany
| | - Annika Gross
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany
| | - Raymond Kwan
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, and the VA Ann Arbor Health Care System, Ann Arbor, MI, USA
| | - Laura P. James
- Arkansas Children's Hospital Research Institute and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Christian Trautwein
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany
| | - M. Bishr Omary
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, and the VA Ann Arbor Health Care System, Ann Arbor, MI, USA
| | - Pavel Strnad
- IZKF and Department of Internal Medicine III, University Hospital Aachen, Germany,Department of Internal Medicine I, University Hospital Ulm, Ulm Germany,To whom correspondence should be addressed. Corresponding author: Pavel Strnad, Department of Internal Medicine III and IZKF, University Hospital Aachen, Pauwelsstraße 30, D-52074 Aachen, Tel.: +49(241) 80-35324, Fax: +49(241) 80-82455,
| |
Collapse
|
35
|
Usachov V, Urban TJ, Fontana RJ, Gross A, Iyer S, Omary MB, Strnad P. Prevalence of genetic variants of keratins 8 and 18 in patients with drug-induced liver injury. BMC Med 2015; 13:196. [PMID: 26286715 PMCID: PMC4545365 DOI: 10.1186/s12916-015-0418-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 07/03/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Keratin 8 and 18 (K8/K18) cytoskeletal proteins protect hepatocytes from undergoing apoptosis and their mutations predispose to adverse outcomes in acute liver failure (ALF). All known K8/K18 variants occur at relatively non-conserved residues and do not cause keratin cytoskeleton reorganization, whereas epidermal keratin-conserved residue mutations disrupt the keratin cytoskeleton and cause severe skin disease. The aim of our study was to identify keratin variants in idiosyncratic drug-induced liver injury (DILI). METHODS Genomic DNA was isolated from 800 patients enrolled in an ongoing US multicenter study, with DILI attributed to a wide range of drugs. Specific K8/K18 exonic regions were PCR-amplified and screened by denaturing HPLC followed by DNA sequencing. The functional impact of keratin variants was assessed using cell transfection and immune staining. RESULTS Heterozygous and compound amino acid-altering K8/K18 variants were identified in 86 DILI patients and non-coding variants in 15 subjects. Five novel amino acid-altering (K8 Lys393Arg, K8 Ala351Val, K8 Ala358Val, K8 Ile346Val, K18 Asp89His) and two non-coding variants were observed. Several variants segregated with specific ethnic backgrounds but were found at similar frequencies in DILI subjects and ethnically matched population controls. Notably, variants in highly conserved residues of K8 Lys393Arg (ezetimibe/simvastatin-related) and K18 Asp89His (isoniazid-related) were found in patients with fatal DILI. These novel variants also led to keratin network disruption in transfected cells. CONCLUSIONS Novel K8/K18 cytoskeleton-disrupting variants were identified in two patients and segregated with fatal DILI. Other non-cytoskeleton-disrupting keratin variants did not preferentially associate with DILI.
Collapse
Affiliation(s)
- Valentyn Usachov
- Department of Internal Medicine III and IZKF, University Hospital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany. .,Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany.
| | - Thomas J Urban
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, UNC Eshelman School of Pharmacy, UNC Hamner Institute for Drug Safety Sciences, University of North Carolina, Chapel Hill, NC, USA.
| | - Robert J Fontana
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA.
| | - Annika Gross
- Department of Internal Medicine III and IZKF, University Hospital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| | - Sapna Iyer
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Pavel Strnad
- Department of Internal Medicine III and IZKF, University Hospital Aachen, RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany.
| | | |
Collapse
|
36
|
Tumor necrosis factor-α confers cardioprotection through ectopic expression of keratins K8 and K18. Nat Med 2015; 21:1076-84. [PMID: 26280121 DOI: 10.1038/nm.3925] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/15/2015] [Indexed: 12/18/2022]
Abstract
Tumor necrosis factor-α (TNF-α), one of the major stress-induced proinflammatory cytokines, is upregulated in the heart after tissue injury, and its sustained expression can contribute to the development of heart failure. Whether TNF-α also exerts cytoprotective effects in heart failure is not known. Here we provide evidence for a cardioprotective function of TNF-α in a genetic heart failure model, desmin-deficient mice. The cardioprotective effects of TNF-α are a consequence of nuclear factor-κB (NF-κB)-mediated ectopic expression in cardiomyocytes of keratin 8 (K8) and keratin 18 (K18), two epithelial-specific intermediate filament proteins. In cardiomyocytes, K8 and K18 (K8/K18) formed an alternative cytoskeletal network that localized mainly at intercalated discs (IDs) and conferred cardioprotection by maintaining normal ID structure and mitochondrial integrity and function. Ectopic induction of K8/K18 expression in cardiomyocytes also occurred in other genetic and experimental models of heart failure. Loss of the K8/K18 network resulted in a maladaptive cardiac phenotype following transverse aortic constriction. In human failing myocardium, where TNF-α expression is upregulated, K8/K18 were also ectopically expressed and localized primarily at IDs, which did not contain detectable amounts of desmin. Thus, TNF-α- and NF-κB-mediated formation of an alternative, stress-induced intermediate filament cytoskeleton has cardioprotective function in mice and potentially in humans.
Collapse
|
37
|
Helenius TO, Misiorek JO, Nyström JH, Fortelius LE, Habtezion A, Liao J, Asghar MN, Zhang H, Azhar S, Omary MB, Toivola DM. Keratin 8 absence down-regulates colonocyte HMGCS2 and modulates colonic ketogenesis and energy metabolism. Mol Biol Cell 2015; 26:2298-310. [PMID: 25904331 PMCID: PMC4462946 DOI: 10.1091/mbc.e14-02-0736] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/15/2015] [Indexed: 12/22/2022] Open
Abstract
Absence of colonic keratin 8 causes intestinal inflammation and decreased levels of the ketogenic enzyme HMGCS2. Upstream, the butyrate transporter MCT1 is decreased, leading to increased luminal butyrate. Ketogenic conditions fail to induce HMGCS2 in the keratin 8–knockout colon, suggesting a role for keratins in colonocyte energy homeostasis. Simple-type epithelial keratins are intermediate filament proteins important for mechanical stability and stress protection. Keratin mutations predispose to human liver disorders, whereas their roles in intestinal diseases are unclear. Absence of keratin 8 (K8) in mice leads to colitis, decreased Na/Cl uptake, protein mistargeting, and longer crypts, suggesting that keratins contribute to intestinal homeostasis. We describe the rate-limiting enzyme of the ketogenic energy metabolism pathway, mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), as a major down-regulated protein in the K8-knockout (K8−/−) colon. K8 absence leads to decreased quantity and activity of HMGCS2, and the down-regulation is not dependent on the inflammatory state, since HMGCS2 is not decreased in dextran sulfate sodium-induced colitis. Peroxisome proliferator–activated receptor α, a transcriptional activator of HMGCS2, is similarly down-regulated. Ketogenic conditions—starvation or ketogenic diet—increase K8+/+ HMGCS2, whereas this response is blunted in the K8−/− colon. Microbiota-produced short-chain fatty acids (SCFAs), substrates in the colonic ketone body pathway, are increased in stool, which correlates with decreased levels of their main transporter, monocarboxylate transporter 1 (MCT1). Microbial populations, including the main SCFA-butyrate producers in the colon, were not altered in the K8−/−. In summary, the regulation of the SCFA-MCT1-HMGCS2 axis is disrupted in K8−/− colonocytes, suggesting a role for keratins in colonocyte energy metabolism and homeostasis.
Collapse
Affiliation(s)
- Terhi O Helenius
- Cell Biology/Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Julia O Misiorek
- Cell Biology/Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Joel H Nyström
- Cell Biology/Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Lina E Fortelius
- Cell Biology/Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Palo Alto, CA 94305
| | | | - M Nadeem Asghar
- Cell Biology/Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Haiyan Zhang
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, and Stanford University School of Medicine, Palo Alto, CA 94304
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, and Stanford University School of Medicine, Palo Alto, CA 94304
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109 VA Ann Arbor Health Care System, Ann Arbor, MI 48105
| | - Diana M Toivola
- Cell Biology/Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| |
Collapse
|
38
|
Piwil2 inhibits keratin 8 degradation through promoting p38-induced phosphorylation to resist Fas-mediated apoptosis. Mol Cell Biol 2014; 34:3928-38. [PMID: 25113562 DOI: 10.1128/mcb.00745-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The piwi-like 2 (piwil2) gene is widely expressed in tumors and protects cells from apoptosis induced by a variety of stress stimuli. However, the role of Piwil2 in Fas-mediated apoptosis remains unknown. Here, we present evidence that Piwil2 inhibits Fas-mediated apoptosis. By a bacterial two-hybrid screening, we identify a new Piwil2-interacting partner, keratin 8 (K8), a major intermediate filament protein protecting the cell from Fas-mediated apoptosis. Our results show that Piwil2 binds to K8 and p38 through its PIWI domain and forms a Piwil2/K8/P38 triple protein-protein complex. Thus, Piwil2 increases the phosphorylation level of K8 Ser-73 and then inhibits ubiquitin-mediated degradation of K8. As a result, the knockdown of Piwil2 increases the Fas protein level at the membrane. In addition to our previous finding that Piwil2 inhibits the expression of p53 through the Src/STAT3 pathway, here we demonstrate that Piwil2 represses p53 phosphorylation through p38. Our present study indicates that Piwil2 plays a role in Fas-mediated apoptosis for the first time and also can affect p53 phosphorylation in tumor cells, revealing a novel mechanism of Piwil2 in apoptosis, and supports that Piwil2 plays an active role in tumorigenesis.
Collapse
|
39
|
|
40
|
Alvarado DM, Coulombe PA. Directed expression of a chimeric type II keratin partially rescues keratin 5-null mice. J Biol Chem 2014; 289:19435-47. [PMID: 24867950 DOI: 10.1074/jbc.m114.553867] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The crucial role of structural support fulfilled by keratin intermediate filaments (IFs) in surface epithelia likely requires that they be organized into cross-linked networks. For IFs comprised of keratins 5 and 14 (K5 and K14), which occur in basal keratinocytes of the epidermis, formation of cross-linked bundles is, in part, self-driven through cis-acting determinants. Here, we targeted the expression of a bundling-competent KRT5/KRT8 chimeric cDNA (KRT8bc) or bundling-deficient wild type KRT8 as a control to the epidermal basal layer of Krt5-null mice to assess the functional importance of keratin IF self-organization in vivo. Such targeted expression of K8bc rescued Krt5-null mice with a 47% frequency, whereas K8 completely failed to do so. This outcome correlated with lower than expected levels of K8bc and especially K8 mRNA and protein in the epidermis of E18.5 replacement embryos. Ex vivo culture of embryonic skin keratinocytes confirmed the ability of K8bc to form IFs in the absence of K5. Additionally, electron microscopy analysis of E18.5 embryonic skin revealed that the striking defects observed in keratin IF bundling, cytoarchitecture, and mitochondria are partially restored by K8bc expression. As young adults, viable KRT8bc replacement mice develop alopecia and chronic skin lesions, indicating that the skin epithelia are not completely normal. These findings are consistent with a contribution of self-mediated organization of keratin IFs to structural support and cytoarchitecture in basal layer keratinocytes of the epidermis and underscore the importance of context-dependent regulation for keratin genes and proteins in vivo.
Collapse
Affiliation(s)
- David M Alvarado
- From the Training Program in Cellular and Molecular Medicine and Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Pierre A Coulombe
- From the Training Program in Cellular and Molecular Medicine and Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland 21205Departments of Biological Chemistry and Dermatology, School of Medicine and
| |
Collapse
|
41
|
Abstract
Neurons, perhaps more than any other cell type, depend on mitochondrial trafficking for their survival. Recent studies have elucidated a motor/adaptor complex on the mitochondrial surface that is shared between neurons and other animal cells. In addition to kinesin and dynein, this complex contains the proteins Miro (also called RhoT1/2) and milton (also called TRAK1/2) and is responsible for much, although not necessarily all, mitochondrial movement. Elucidation of the complex has permitted inroads for understanding how this movement is regulated by a variety of intracellular signals, although many mysteries remain. Regulating mitochondrial movement can match energy demand to energy supply throughout the extraordinary architecture of these cells and can control the clearance and replenishing of mitochondria in the periphery. Because the extended axons of neurons contain uniformly polarized microtubules, they have been useful for studying mitochondrial motility in conjunction with biochemical assays in many cell types.
Collapse
Affiliation(s)
- Thomas L Schwarz
- F.M. Kirby Neurobiology Center, Children's Hospital Boston, and Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
42
|
Tao GZ, Lehwald N, Jang KY, Baek J, Xu B, Omary MB, Sylvester KG. Wnt/β-catenin signaling protects mouse liver against oxidative stress-induced apoptosis through the inhibition of forkhead transcription factor FoxO3. J Biol Chem 2013; 288:17214-24. [PMID: 23620592 DOI: 10.1074/jbc.m112.445965] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Numerous liver diseases are associated with extensive oxidative tissue damage. It is well established that Wnt/β-catenin signaling directs multiple hepatocellular processes, including development, proliferation, regeneration, nutrient homeostasis, and carcinogenesis. It remains unexplored whether Wnt/β-catenin signaling provides hepatocyte protection against hepatotoxin-induced apoptosis. Conditional, liver-specific β-catenin knockdown (KD) mice and their wild-type littermates were challenged by feeding with a hepatotoxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet to induce chronic oxidative liver injury. Following the DDC diet, mice with β-catenin-deficient hepatocytes demonstrate increased liver injury, indicating an important role of β-catenin signaling for liver protection against oxidative stress. This finding was further confirmed in AML12 hepatocytes with β-catenin signaling manipulation in vitro using paraquat, a known oxidative stress inducer. Immunofluorescence staining revealed an intense nuclear FoxO3 staining in β-catenin-deficient livers, suggesting active FoxO3 signaling in response to DDC-induced liver injury when compared with wild-type controls. Consistently, FoxO3 target genes p27 and Bim were significantly induced in β-catenin KD livers. Conversely, SGK1, a β-catenin target gene, was significantly impaired in β-catenin KD hepatocytes that failed to inactivate FoxO3. Furthermore, shRNA-mediated deletion of FoxO3 increased hepatocyte resistance to oxidative stress-induced apoptosis, confirming a proapoptotic role of FoxO3 in the stressed liver. Our findings suggest that Wnt/β-catenin signaling is required for hepatocyte protection against oxidative stress-induced apoptosis. The inhibition of FoxO through its phosphorylation by β-catenin-induced SGK1 expression reduces the apoptotic function of FoxO3, resulting in increased hepatocyte survival. These findings have relevance for future therapies directed at hepatocyte protection, regeneration, and anti-cancer treatment.
Collapse
Affiliation(s)
- Guo-Zhong Tao
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305-5148, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Proteomics identifies molecular networks affected by tetradecylthioacetic acid and fish oil supplemented diets. J Proteomics 2013; 84:61-77. [PMID: 23568020 DOI: 10.1016/j.jprot.2013.03.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 03/08/2013] [Accepted: 03/11/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Fish oil (FO) and tetradecylthioacetic acid (TTA) - a synthetic modified fatty acid have beneficial effects in regulating lipid metabolism. In order to dissect the mechanisms underlying the molecular action of those two fatty acids we have investigated the changes in mitochondrial protein expression in a long-term study (50weeks) in male Wistar rats fed 5 different diets. The diets were as follows: low fat diet; high fat diet; and three diets that combined high fat diet with fish oil, TTA or combination of those two as food supplements. We used two different proteomics techniques: a protein centric based on 2D gel electrophoresis and mass spectrometry, and LC-MS(E) based peptide centric approach. As a result we provide evidence that fish oil and TTA modulate mitochondrial metabolism in a synergistic manner yet the effects of TTA are much more dramatic. We demonstrate that fatty acid metabolism; lipid oxidation, amino acid metabolism and oxidative phosphorylation pathways are involved in fish oil and TTA action. Evidence for the involvement of PPAR mediated signalling is provided. Additionally we postulate that down regulation of components of complexes I and II contributes to the strong antioxidant properties of TTA. BIOLOGICAL SIGNIFICANCE This study for the first time explores the effect of fish oil and TTA - tetradecyl-thioacetic acid and the combination of those two as diet supplements on mitochondria metabolism in a comprehensive and systematic manner. We show that fish oil and TTA modulate mitochondrial metabolism in a synergistic manner yet the effects of TTA are much more dramatic. We demonstrate in a large scale that fatty acid metabolism and lipid oxidation are affected by fish oil and TTA, a phenomenon already known from more directed molecular biology studies. Our approach, however, shows additionally that amino acid metabolism and oxidative phosphorylation pathways are also strongly affected by TTA and also to some extent by fish oil administration. Strong evidence for the involvement of PPAR mediated signalling is provided linking the different metabolic effects. The global and systematic viewpoint of this study compiles many of the known phenomena related to the effects of fish oil and fatty acids giving a solid foundation for further exploratory and more directed studies of the mechanisms behind the beneficial and detrimental effects of fish oil and TTA diet supplementation. This work is already a second article in a series of studies conducted using this model of dietary intervention. In the previous study (Vigerust et al., [21]) the effects of fish oil and TTA on the plasma lipids and cholesterol levels as well as key metabolic enzymes in the liver have been studied. In an ongoing study more work is being done to explore in detail for example the link between the down regulation of the components of the respiratory chain (observed in this study) and the strong antioxidant effects of TTA. The reference diet in this study has been designed to mimic an unhealthy - high fat diet that is thought to contribute to the development of metabolic syndrome - a condition that is strongly associated with diabetes, obesity and heart failure. Fish oil and TTA are known to have beneficial effects for the fatty acid metabolism and have been shown to alleviate some of the symptoms of the metabolic syndrome. To date very little is known about the molecular mechanisms behind these beneficial effects and the potential pitfalls of the consumption of those two compounds. Only studies of each compound separately and using only small scale molecular biology approaches have been carried out. The results of this work provide an excellent starting point for further studies that will help to understand the metabolic effects of fish oil and TTA and will hopefully help to design dietary programs directed towards reduction of the prevalence of metabolic syndrome and associated diseases.
Collapse
|
44
|
Biological features of core networks that result from a high-fat diet in hepatic and pulmonary tissues in mammary tumour-bearing, obesity-resistant mice. Br J Nutr 2012; 110:241-55. [DOI: 10.1017/s0007114512004965] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We previously demonstrated that the chronic consumption of a high-fat diet (HFD) promotes lung and liver metastases of 4T1 mammary carcinoma cells in obesity-resistant BALB/c mice. To examine early transcriptional responses to tumour progression in the liver and lungs of HFD-fed mice, 4-week-old female BALB/c mice were divided into four groups: sham-injected, control diet (CD)-fed; sham-injected, HFD-fed (SH); 4T1 cell-injected, CD-fed (TC); 4T1 cell-injected, HFD-fed (TH). Following 16 weeks of either a CD or HFD, 4T1 cells were injected into the mammary fat pads of mice in the TC and TH groups and all mice were continuously fed identical diets. At 14 d post-injection, RNA was isolated from hepatic and pulmonary tissues for microarray analysis of mRNA expression. Functional annotation and core network analyses were conducted for the TH/SH Unique gene set. Inflammation in hepatic tissues and cell mitosis in pulmonary tissues were the most significant biological functions in the TH/SH Unique gene set. The biological core networks of the hepatic TH/SH Unique gene set were characterised as those genes involved in the activation of acute inflammatory responses (Orm1, Lbp, Hp and Cfb), disordered lipid metabolism and deregulated cell cycle progression. Networks of the pulmonary Unique gene set displayed the deregulation of cell cycle progression (Cdc20, Cdk1 and Bub1b). These HFD-influenced alterations may have led to favourable conditions for the formation of both pro-inflammatory and pro-mitotic microenvironments in the target organs that promote immune cell infiltration and differentiation, as well as the infiltration and proliferation of metastatic tumour cells.
Collapse
|
45
|
Lehwald N, Tao GZ, Jang KY, Papandreou I, Liu B, Liu B, Pysz MA, Willmann JK, Knoefel WT, Denko NC, Sylvester KG. β-Catenin regulates hepatic mitochondrial function and energy balance in mice. Gastroenterology 2012; 143:754-764. [PMID: 22684045 PMCID: PMC12045480 DOI: 10.1053/j.gastro.2012.05.048] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Revised: 05/07/2012] [Accepted: 05/29/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Wnt signaling regulates hepatic function and nutrient homeostasis. However, little is known about the roles of β-catenin in cellular respiration or mitochondria of hepatocytes. METHODS We investigated β-catenin's role in the metabolic function of hepatocytes under homeostatic conditions and in response to metabolic stress using mice with hepatocyte-specific deletion of β-catenin and their wild-type littermates, given either saline (sham) or ethanol (as a model of binge drinking and acute ethanol intoxication). RESULTS Under homeostatic conditions, β-catenin-deficient hepatocytes demonstrated mitochondrial dysfunctions that included impairments to the tricarboxylic acid cycle and oxidative phosphorylation (OXPHOS) and decreased production of adenosine triphosphate (ATP). There was no evidence for redox imbalance or oxidative cellular injury in the absence of metabolic stress. In mice with β-catenin-deficient hepatocytes, ethanol intoxication led to significant redox imbalance in the hepatocytes and further deterioration in mitochondrial function that included reduced OXPHOS, fatty acid oxidation (FAO), and ATP production. Ethanol feeding significantly increased liver steatosis and oxidative damage, compared with wild-type mice, and disrupted the ratio of nicotinamide adenine dinucleotide. β-catenin-deficient hepatocytes also had showed disrupted signaling of Sirt1/peroxisome proliferator-activated receptor-α signaling. CONCLUSIONS β-catenin has an important role in the maintenance of mitochondrial homeostasis, regulating ATP production via the tricarboxylic acid cycle, OXPHOS, and fatty acid oxidation; β-catenin function in these systems is compromised under conditions of nutrient oxidative stress. Reagents that alter Wnt-β-catenin signaling might be developed as a useful new therapeutic strategy for treatment of liver disease.
Collapse
MESH Headings
- Adenosine Triphosphate
- Animals
- Citric Acid Cycle
- Disease Models, Animal
- Energy Metabolism/drug effects
- Ethanol/toxicity
- Fatty Acids/metabolism
- Fatty Liver, Alcoholic/etiology
- Fatty Liver, Alcoholic/metabolism
- Fatty Liver, Alcoholic/pathology
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Homeostasis
- Lipid Peroxidation
- Membrane Potential, Mitochondrial
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Liver/drug effects
- Mitochondria, Liver/metabolism
- Mitochondria, Liver/pathology
- Oxidation-Reduction
- Oxidative Phosphorylation
- Oxidative Stress
- Time Factors
- Wnt Signaling Pathway/drug effects
- beta Catenin/deficiency
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Nadja Lehwald
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California; Department of General, Visceral, and Pediatric Surgery, School of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - Guo-Zhong Tao
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Kyu Yun Jang
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California; Department of Pathology and Research Institute of Clinical Medicine, Chonbuk National University Medical School, South Korea
| | - Ioanna Papandreou
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Bowen Liu
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Bo Liu
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Marybeth A Pysz
- Department of Radiology, Molecular Imaging Program, Stanford University School of Medicine, Stanford, California
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program, Stanford University School of Medicine, Stanford, California
| | - Wolfram T Knoefel
- Department of General, Visceral, and Pediatric Surgery, School of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - Nicholas C Denko
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Karl G Sylvester
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California; The Lucile Packard Children's Hospital, Stanford, California.
| |
Collapse
|
46
|
Singla A, Moons DS, Snider NT, Wagenmaker ER, Jayasundera VB, Omary MB. Oxidative stress, Nrf2 and keratin up-regulation associate with Mallory-Denk body formation in mouse erythropoietic protoporphyria. Hepatology 2012; 56:322-31. [PMID: 22334478 PMCID: PMC3389581 DOI: 10.1002/hep.25664] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 01/27/2012] [Indexed: 01/02/2023]
Abstract
UNLABELLED Mallory-Denk bodies (MDBs) are hepatocyte inclusions commonly seen in steatohepatitis. They are induced in mice by feeding 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) for 12 weeks, which also causes porphyrin accumulation. Erythropoietic protoporphyria (EPP) is caused by mutations in ferrochelatase (fch), and a fraction of EPP patients develop liver disease that is phenocopied in Fech(m1Pas) mutant (fch/fch) mice, which have an inactivating fch mutation. fch/fch mice develop spontaneous MDBs, but the molecular factors involved in their formation and whether they relate to DDC-induced MDBs are unknown. We tested the hypothesis that fch mutation creates a molecular milieu that mimics experimental drug-induced MDBs. In 13- and 20-week-old fch/fch mice, serum alkaline phosphatase, alanine aminotransferase, and bile acids were increased. The 13-week-old fch/fch mice did not develop histologically evident MDBs but manifested biochemical alterations required for MDB formation, including increased transglutaminase-2 and keratin overexpression, with a greater keratin 8 (K8)-to-keratin 18 (K18) ratio, which are critical for drug-induced MDB formation. In 20-week-old fch/fch mice, spontaneous MDBs were readily detected histologically and biochemically. Short-term (3-week) DDC feeding markedly induced MDB formation in 20-week-old fch/fch mice. Under basal conditions, old fch/fch mice had significant alterations in mitochondrial oxidative-stress markers, including increased protein oxidation, decreased proteasomal activity, reduced adenosine triphosphate content, and Nrf2 (redox sensitive transcription factor) up-regulation. Nrf2 knockdown in HepG2 cells down-regulated K8, but not K18. CONCLUSION Fch/fch mice develop age-associated spontaneous MDBs, with a marked propensity for rapid MDB formation upon exposure to DDC, and therefore provide a genetic model for MDB formation. Inclusion formation in the fch/fch mice involves oxidative stress which, together with Nrf2-mediated increase in K8, promotes MDB formation.
Collapse
Affiliation(s)
- Amika Singla
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-5622
| | - David S. Moons
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-5622
| | - Natasha T. Snider
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-5622
| | - Elizabeth R. Wagenmaker
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-5622
| | - V. Bernadene Jayasundera
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-5622
| | - M. Bishr Omary
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-5622,Department of Medicine, University of Michigan Medical School, Ann Arbor, MI 48109-5622,To whom correspondence should be addressed: Bishr Omary, University of Michigan Medical School, Department of Molecular & Integrative Physiology, 7744 Medical Science Building II, 1137 Catherine St., Ann Arbor, MI 48109, Phone: 734-764-4376, Fax: 734-936-8813,
| |
Collapse
|
47
|
Gentil BJ, Cooper L. Molecular basis of axonal dysfunction and traffic impairments in CMT. Brain Res Bull 2012; 88:444-53. [PMID: 22595495 DOI: 10.1016/j.brainresbull.2012.05.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 05/01/2012] [Accepted: 05/04/2012] [Indexed: 12/17/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) is one of the most common inherited neurological disorders. It comprises a group of diseases caused by mutations in genes involved in Schwann cells homeostasis and neuronal function that affect the peripheral nerves. So far mutations in more than 33 genes have been identified causing either the demyelinating form (CMT1) or the axonal form (CMT2). Genes involving a large variety of unrelated functions may lead to the same phenotype when mutated. Our review will focus on the common link between genes causing axonal phenotypes like MFN2, KIF1B, DYNC1H1, Rab7, TRPV4, ARSs, NEFL, HSPB1, MPZ, and HSPB8. While KIF1B and DYNC1H1, two genes coding for molecular motors, are directly linked to axonal transport, the involvement of the other CMT2-causing genes in this function is less obvious. However, the last years have seen a growing list of evidence demonstrating that intracellular trafficking and mitochondrial dynamics might be dysfunctional in CMT2, and these mechanisms might present a common link between dissimilar CMT2-causing genes. The involvement of impaired transport in the pathogenesis of other rare neurological diseases or recessive CMT2 is also discussed.
Collapse
Affiliation(s)
- Benoit J Gentil
- Department of Neurology/Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4 Canada.
| | | |
Collapse
|
48
|
Lincet H, Guével B, Pineau C, Allouche S, Lemoisson E, Poulain L, Gauduchon P. Comparative 2D-DIGE proteomic analysis of ovarian carcinoma cells: Toward a reorientation of biosynthesis pathways associated with acquired platinum resistance. J Proteomics 2012; 75:1157-69. [DOI: 10.1016/j.jprot.2011.10.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 09/29/2011] [Accepted: 10/27/2011] [Indexed: 01/08/2023]
|
49
|
Nekrasova OE, Mendez MG, Chernoivanenko IS, Tyurin-Kuzmin PA, Kuczmarski ER, Gelfand VI, Goldman RD, Minin AA. Vimentin intermediate filaments modulate the motility of mitochondria. Mol Biol Cell 2011; 22:2282-9. [PMID: 21562225 PMCID: PMC3128530 DOI: 10.1091/mbc.e10-09-0766] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 04/19/2011] [Accepted: 05/05/2011] [Indexed: 12/17/2022] Open
Abstract
Interactions with vimentin intermediate filaments (VimIFs) affect the motility, distribution, and anchorage of mitochondria. In cells lacking VimIFs or in which VimIF organization is disrupted, the motility of mitochondria is increased relative to control cells that express normal VimIF networks. Expression of wild-type VimIF in vimentin-null cells causes mitochondrial motility to return to normal (slower) rates. In contrast, expressing vimentin with mutations in the mid-region of the N-terminal non-α-helical domain (deletions of residues 41-96 or 45-70, or substitution of Pro-57 with Arg) did not inhibit mitochondrial motility even though these mutants retain their ability to assemble into VimIFs in vivo. It was also found that a vimentin peptide consisting of residues 41-94 localizes to mitochondria. Taken together, these data suggest that VimIFs bind directly or indirectly to mitochondria and anchor them within the cytoplasm.
Collapse
Affiliation(s)
- Oxana E. Nekrasova
- Group of Cell Biology, Institute of Protein Research, Russian Academy of Sciences, Moscow 119988, Russia
| | - Melissa G. Mendez
- Department of Cell and Molecular Biology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611
| | - Ivan S. Chernoivanenko
- Group of Cell Biology, Institute of Protein Research, Russian Academy of Sciences, Moscow 119988, Russia
- Koltsov's Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Pyotr A. Tyurin-Kuzmin
- Group of Cell Biology, Institute of Protein Research, Russian Academy of Sciences, Moscow 119988, Russia
| | - Edward R. Kuczmarski
- Department of Cell and Molecular Biology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611
| | - Vladimir I. Gelfand
- Department of Cell and Molecular Biology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611
| | - Robert D. Goldman
- Department of Cell and Molecular Biology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611
| | - Alexander A. Minin
- Group of Cell Biology, Institute of Protein Research, Russian Academy of Sciences, Moscow 119988, Russia
| |
Collapse
|
50
|
Jayakumar S, Guillot S, Argo C, Redick J, Caldwell S. Ultrastructural findings in human nonalcoholic steatohepatitis. Expert Rev Gastroenterol Hepatol 2011; 5:141-5. [PMID: 21476907 DOI: 10.1586/egh.11.9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|